DOI: 10.17344/acsi.2015.1668 Acta Chim. Slov. 2016, 63, 227-240 227 Scientific paper New Approaches for the Synthesis, Cytotoxicity and Toxicity of Heterocyclic Compounds Derived from 2-Cyanomethylbenzo[c]imidazole Rafat M. Mohareb,1* Abeer A. Mohamed2 and Amira E. M. Abdallah3 1 Department of Chemistry, Faculty of Science, Cairo University, Giza, A. R. Egypt 2 National Organization for Research & Control of Biologicals, Giza, Egypt 3 Department of Chemistry, Faculty of Science, Helwan University, Cario, Egypt * Corresponding author: E-mail: raafat_mohareb@yahoo.com Received: 04-05-2015 Abstract The reaction of ethyl cyanoacetate with o-phenylenediamine gave the 2-cyanomethylbenzo[c]imidazole (1). The latter compound was used as the key starting material to synthesise biologically active heterocyclic derivatives. Thus, the reaction of 1 with cyclohexanone and either of benzaldehyde, 4-methoxybenzaldehyde or 4-chlorobenzaldehyde gave the annulated derivatives 2a-c, respectively. The antitumor evaluations of the newly synthesized products against the three cancer cell lines MCF-7 (breast adeno-carcinoma), NCI-H460 (non-small cell lung cancer) and SF-268 (CNS cancer) showed that compounds 2b, 6, 11b, 11c, 12b, 16a, 16b and 18a exhibited optimal cytotoxic effect against cancer cell lines, with IC50 values in the nM range. Bioactive compounds are often toxic to shrimp larvae. Thus, in order to monitor these chemicals in vivo lethality to shrimp larvae (Artemia salina), Brine-Shrimp Lethality Assay was used. Compounds 11b, 12b and 16b showed no toxicity against the tested organisms. Keywords: benzimidazole, thiophene, thiazole, synthesis, anti-tumor, toxicity 1. Introduction In recent years benzimidazole derivatives have provided a large number of biologically active compounds that have been intensively used in medicinal chemistry as drugs. They are structural isosteres of naturally occurring nucleotides, which allow them to interact easily with the biopolymers of the living systems and various kinds of biological activities have been obtained. Some 2-aminobenzimidazoles display an appreciable antimicrobial effect. Their corresponding carbamate derivatives have been synthesized for their significant in vivo antifilarial activity.1 Concerning the high affinity that they display towards a variety of enzymes and protein receptors, they could be considered as pivotal structures in drug design.2 Optimization of benzimidazole-based structures has resulted in marketed drugs, e.g. Omeprazole3 and Pimobendan4 that are therapeutically useful in the management of peptic ulcer and congestive heart failure, respectively. Many derivatives of benzimidazoles are well known for their antimicrobial,5-10 anthelmintic,11 antiviral,12-16 and antifungal17,18 activities. Since 1985 benzimidazole containing compounds have been reported as well known anticancer agents.19-25 The role of mammalian DNA topoisomerases as molecular targets for anticancer drugs has been recognized. Some benzimidazoles have been reported as topoisomerase inhibitors, e.g. Hoechst 33258 and Hoechst 33342 (Fig. 1).26,27 As the extension of this work, head to head bis-benzimidazole compounds proved high efficacy as DNA binders.28 Some widely used anticancer drugs such as RAF265 (CHIR-265; Novartis Pharmaceuticals, Basel, Switzerland) and AZD6244 (ARRY-142886; AstraZene-ca, London, England) are known to contain benzimida-zole moiety. RAF265 resulted in a reduction in tumor cell growth and in tumor cell apoptosis.29 Mohareb et al.: New Approaches for the Synthesis, Cytotoxicity 228 Acta Chim. Slov. 2016, 63, 227-240 Hoechst 33342 Fig. 1. Examples of topoisomerase inhibitors containing benzimidazole nucleus. 2. Results and Discussion 2. 1. Chemistry The 2-cyanomethylbenzo[c]imidazole (1) obtained from the reaction of ethyl cyanoacetate with o-phenylene- diamine was used as the key starting material to synthe-sise biologically active heterocyclic derivatives. Thus, the reaction of 1 with cyclohexanone and any of benzaldehyde, 4-methoxybenzaldehyde or 4-chlorobenzaldehyde gave the annulated derivatives 2a-c, respectively. The analy- Scheme 1. Mohareb et al.: New Approaches for the Synthesis, Cytotoxicity Acta Chim. Slov. 2016, 63, 227-240 229 tical and spectral data of 2a-c were consistent with their respective structures. Thus, the NMR spectrum of 2a (as an example) showed 2.49-2.88 (CH2-cyclohexanone), 7.13-8.01 (m, 9H, C6H4 C6H5). Moreover, the 13C NMR data revealed 38.6, 39.0, 40.2, 40.6 (4 x CH2-cyclohexa-none), 116.2 (CN), 120.3, 122.8, 124.9, 127.8, 128.0, 129.8, 131.2, 132.6, 134.5, 134.8, 144.3, 146.8, 150.4 (2 x C6H4, pyridine C), 164.8 (C=N). The reaction of 1 with acetoacetanilide gave the ben-zo[c]pyrazolo[3,2-a]pyridine derivative 3. The latter compound reacted with benzene diazonium chloride to give the phenylazo derivative 4 (Scheme 1). On the other hand, the reaction of 1 with acetic acid/acetic anhydride mixture gave the N-acetyl derivative 5. Compound 5 readily underwent bromination when treated with bromine in acetic acid solution at 60 oC to give the N-a-bromoacetylbenzo[c]imi-dazole derivative 6. The latter compound as a-bromocar-bonyl compound showed interesting chemical reactivity when treated with some chemical reagents. Thus, the reaction of 6 with potassium cyanide gave the benzo[c]imida-zo[2,3-fc]pyridine derivative 7. On the other hand, the reaction of 6 with hydrazine hydrate afforded the hydrazine de- Scheme 2. Mohareb et al.: New Approaches for the Synthesis, Cytotoxicity 230 Acta Chim. Slov. 2016, 63, 227-240 230 rivative 8. Compound 6 reacted with thiourea in ethanol to give the thiazole derivative 9 (Scheme 2). Recently, our research group was involved in a comprehensive program involving the reaction of active methylene reagents with phenylisothiocyanate in basic (KOH) dimethylformamide to form the intermediate potassium sulphide salt. The latter undergoes heterocycliza-tion when reacted with a-halocarbonyl compounds to give either thiophene or thiazole derivatives depending on the nature of the a-halocarbonyl compound and the reaction conditions.30-32 Thus, the reaction of 5 with phenylisothiocyanate in DMF/KOH solution gave the intermediate potassium sulphide salt 10. The latter intermediate underwent heterocyclization when reacted with any of a-chloroacetone, ethyl chloroacetate or a-bromoacetophe-none to give the thiophene derivatives 11a-c, respectively. The analytical and spectral data of 11a-c are consistent with their respective structures. Scheme 2. Mohareb et al.: New Approaches for the Synthesis, Cytotoxicity Acta Chim. Slov. 2016, 63, 227-240 231 The reaction of 5 with either benzaldehyde or sa-licylaldehyde gave the benzylidene derivatives 12a and 12b, respectively. On the other hand, the reaction of 5 with either malononitrile or ethyl cyanoacetate in DMF containing triethylamine gave the 1,5-dihydroben-zo[4,5]imidazo[1,2-a]pyridine derivatives 13a and 13b, respectively (Scheme 3). Compound 5 reacted with acetophenone in an oil bath at 120 °C to give the Knoevenagel condensation pro- duct 14. The latter compound reacted with benzaldehyde to give the benzylidene derivative 15. The reactivity of 5 towards the well-known Gewald's thiophene synthesis was studied to give biologically active thiophene derivatives. Thus, the reaction of 5 with elemental sulfur and either of malononitrile or ethyl cyanoacetate gave the thiophene derivatives 16a and 16b, respectively. On the other hand, the reaction of 5 with benzenediazo-nium chloride in ethanol/sodium hydroxide solution affor- Scheme 2. Mohareb et al.: New Approaches for the Synthesis, Cytotoxicity 232 Acta Chim. Slov. 2016, 63, 227-240 ded the phenylhydrazo derivative 17. Compound 17 underwent the Gewald's thiophene synthesis through the N-acetyl moiety when reacted with elemental sulfur and either of malononitrile or ethyl cyanoacetate in 1,4-dioxane containing triethylamine under reflux to give the thiophene derivatives 18a and 18b, respectively (Scheme 4). 3. Anti-tumor and Normal Cell Line Activity Tests 3. 1. Chemicals Reagents: Fetal bovine serum (FBS) and L-glutami-ne were from Gibco Invitrogen Co. (Scotland, UK). RP-MI-1640 medium was from Cambrex (New Jersey, USA). Dimethyl sulfoxide (DMSO), doxorubicin, penicillin, streptomycin and sulforhodamine B (SRB) were from Sigma Chemical Co. (Saint Louis, USA). 3. 1. 1. Cell Cultures Three human tumor cell lines, MCF-7 (breast adenocarcinoma), NCI-H460 (non-small cell lung cancer), and SF-268 (CNS cancer) were used. MCF-7 was obtained from the European Collection of Cell Cultures (ECACC, Salisbury, UK), NCI-H460, SF-268 and normal fibroblast were grown as monolayer and routinely maintained in RPMI-1640 medium supplemented with 5% heat inactivated FBS, 2 mM glutamine and antibiotics (penicillin 100 U/mL, streptomycin 100 ^g/mL), at 37 oC in a humidified atmosphere containing 5% CO2. Exponentially growing cells were obtained by plating 1.5 x 105 cells/mL for MCF-7 and SF-268 and 0.75 x 104 cells/mL for NCI-H460, followed by 24 h of incubation. The effect of the vehicle solvent (DMSO) on the growth of these cell lines was evaluated in all the experiments by exposing untreated control cells to the maximum concentration (0.5%) of DMSO used in each assay. 3. 1. 2. TUmor Cell Growth Assay The effects of 2a-c to 18a,b on the in vitro growth of human tumor cell lines were evaluated according to the procedure adopted by the National Cancer Institute (NCI, USA) in the zIn vitro Anticancer Drug Discovery Screen' that uses the protein-binding dye sulforhodamine B to assess cell growth.33 Briefly, exponentially, cells growing in Table 1. Effect of newly synthesized compounds on the growth of three human tumor cell lines C d GISo (^ mol L1) Compound MCF-7 NCI-H460 SF-268 WI 38 2a 33.0 ± 1.4 20.8 ± 4.3 20.3 ± 2.8 38.4 ± 2.90 2b 0.8 ± 0.04 0.5 ± 0.02 0.06 ± 0.001 20.0 ± 4.94 2c 22.1 ± 10.4 30.8 ± 10.8 26.1 ± 2.8 28.2 ± 0.8 3 33.6 ± 10.2 40.0 ± 8.6 38.6 ± 8.0 8 >100 4 32.2 ± 3.6 36.3 ± 12.5 40.6 ± 8.8 50.7 ± 8.2 5 22.8 ± 8.30 22.8 ± 4.32 22.8 ± 6.23 44.8 ± 6.0 6 0.01 ± 0.001 0.02 ± 0.004 0.06 ± 0.002 >100 7 28.4 ± 5.8 22.7 ± 8.2 30.4 ± 2.4 18.6 ±4.0 8 23.55 ± 4.06 34.6 ± 12.06 45.41 ± 2.16 >100 9 33.6 ± 8.5 40.3 ± 12.3 30.4 ± 2.8 62.2 ± 2.0 11a 26.4 ± 2.10 12.42 ± 3.01 10.63 ± 2.83 >100 11b 0.81 ± 0.04 0.52 ± 0.04 0.08 ± 0.006 40.0 ± 1.3 11c 1.6 ± 0.4 0.6 ± 0.16 1.8 ± 0.06 22.4 ±1.6 12a 26.2 ± 2.4 28.6 ± 2.8 26.8 ± 8.5 30.2 ± 2.6 12b 0.02 ± 0.001 0.03 ± 0.006 0.06 ± 0.008 > 100 13a 30.22 ± 6.12 28.99 ± 4.70 10.39 ± 6.80 > 100 13b 12.6 ± 2.01 18.6 ± 6.06 30.4 ± 2.36 30.6 ± 10.2 14 12.33 ± 2.16 16.36 ± 2.26 18.20 ± 5.28 55.5 ±8.3 15 30.7 ± 6.2 38.5 ± 6.4 37.5 ± 8.0 66.0 ± 18.4 16a 2.6 ± 2.8 6.6 ± 2.2 5.0 ± 1.81 0.5 ± 5.1 16b 0.06 ± 0.006 0.06 ± 0.006 0.02 ± 0.008 >100 17 38 ± 4. 18 39.03 ± 8.01 22.59 ± 4.01 20.20 ± 8.2 18a 0.08 ± 0.002 0.08 ± 0.003 0.02 ± 0.002 >100 18b 36.0 ± 7.3 26.7 ± 2.8 30.4 ± 2.9 32.6 ± 6.4 Doxorubicin 0.04 ± 0.008 0.09 ± 0.008 0.09 ± 0.007 > 100 Results are given in concentrations that were able to cause 50% of cell growth inhibition (GI50) after a continuous exposure of 48 h and show means ± SEM of three-independent experiments performed in duplicate. Mohareb et al.: New Approaches for the Synthesis, Cytotoxicity ... Acta Chim. Slov. 2016, 63, 227-240 233 96-well plates were then exposed for 48 h to five serial concentrations of each compound, starting from a maximum concentration of 150 pM. Following this exposure period adherent cells were fixed, washed, and stained. The bound stain was solubilized and the absorbance was measured at 492 nm in a plate reader (Bio-Tek Instruments Inc., Power wave XS, Winooski, USA). For each test compound and cell line, a dose-response curve was obtained and the growth inhibition of 50% (GI50), corresponding to the concentration of the compounds that inhibited 50% of the net cell growth was calculated as described elsewhere. Doxorubicin was used as a positive control and tested in the same manner. derivatives 2a-c, it is clear that the cytotoxicity of 2b is higher than those of 2a and 2c. Such high cytotoxicity of 2b is attributed to the presence of the 4-chlorophenylisoquinoline moiety together with the benzimidazole moiety. The high cytotoxicity of 16b relative to 16a is also explained in terms of the presence of the 3-EtO moiety. On the other hand, by considering the (1H-benzo[d]imidazol-1-yl)-2-aminothiop-hene derivatives 18a and 18b it is clear that the presence of the 2-carbonitrile group present in 18a is responsible for its high potency. The bromo-1H-benzo[d]imidazole derivative 6 showed the maximum cytotoxicity effect towards the three cancer cell lines followed by acetyl-1H-benzo[d]imi-dazolhydroxyphenyl derivative 12b. 3. 1. 3. Structure Activity Relationship: From Table 1 it is clear that the benzimidazole moiety was found to be crucial for the cytotoxic effect of the cyclic compounds 2a-c to 18a,b. Compounds 2b, 6, 11b, 11c, 12b, 16a, 16b and 18a exhibited optimal cytotoxic effect against cancer cell lines, with IC50 values in the nM range. Comparing the cytotoxicity of the benzimidazothiophenes 11b and 11c, it is obvious that the cytotoxicity of 11b is higher than that of 11c. The presence of the 2-EtO group in 11b is responsible for its high potency. Considering the 7,8,9,10-tetrahydrobenzo[4,5]imidazo[1,2-fc]isoquinoline 3. 2. Toxicity Bioactive compounds are often toxic to shrimp larvae. Thus, in order to monitor these chemicals, in vivo lethality to shrimp larvae (Artemia salina), Brine-Shrimp Lethality Assay34 was used. Results were analyzed with LC50 program to determine LC50 values and 95% confidence in-tervals.35 Results are given in Table 2 for the compounds which exhibited optimal cytotoxic effect against cancer cell lines, being the eight compounds 2b, 6, 11b, 11c, 12b, 16a, 16b and 18a. The shrimp lethality assay is considered as a useful tool for preliminary assessment of toxicity, and it has Table 2. Toxicity of the most potent compounds against the cancer cell lines Compound Conc. (^g/mL) Mortalitya Toxicity LC50 Upper 95% lim Lower 95% lim 2b 10 100 1000 6 8 10 Very toxic 12.05 6 10 100 1000 1 6 10 Very toxic 18.38 11b 10 100 1000 0 0 2 Non toxic 982.15 11c 10 100 1000 0 5 10 Harmful 420.28 112.23 90.55 12b 10 100 1000 0 1 4 Non toxic 880.42 16a 10 100 1000 2 8 10 Very toxic 14.88 16b 10 100 1000 0 0 5 Non toxic 999.33 18a 10 100 1000 0 6 8 Harmful 22.70 210.59 160.22 a Ten organisms (A. salina) tested for each concentration. Mohareb et al.: New Approaches for the Synthesis, Cytotoxicity ... 234 Acta Chim. Slov. 2016, 63, 227-240 been used for the detection of fungal toxins, plant extract toxicity, heavy metals, cyano bacteria toxins, pesticides, and cytotoxicity testing of dental materials,36 natural and synthetic organic compounds.34 It has also been shown that A. salina toxicity test results have a correlation with rodent and human acute oral toxicity data. Generally, a good correlation was obtained between A. salina toxicity test and the rodent data. Likewise, the predictive screening potential of the aquatic invertebrate tests for acute oral toxicity in man, including A. salina toxicity test, was slightly better than the rat test for test compounds.37 In order to prevent the toxicity results from possible false effects originated from solubility of compounds and DMSO's possible toxicity effect, compounds were prepared by dissolving in DMSO in the suggested DMSO volume ranges. It is clear from Table 2 that 11b, 12b, and 16b showed no toxicity against the tested organisms. On the other hand, 2b, 6 and 16a are very toxic, in addition, 11c and 18a are harmful. 3. 2. 1. Toxicity Method All toxicity tests were 96-h static renewal tests and water quality measurements (dissolved oxygen, pH, temperature, salinity) were taken in the control containers each day. Tests were run in a Revcos Environmental Chamber at 25 °C, 20% salinity, and a 16-h light : 8-h dark cycle. A media change was made every 24 h. Larvae used for all tests were one to two days old and exposed in 600-mL glass beakers containing 400 mL of media with 10 larvae/beaker and three replicates/concentration. Larvae were fed newly hatched Artemia after daily media change. The concentration of each compound was taken in terms 10, 100 and 1000 mg/mL. Adult shrimp toxicity tests were also run to complete the grass shrimp toxicity profile. Adult shrimp (acclimated for two weeks before testing) were exposed in 4-L wide-mouth glass jars containing 2 L of media and 10 shrimp/jar with two replicates/concentration and were run under conditions as described above for larvae.38 4. Experimental All melting points are uncorrected. IR spectra were recorded on KBr discs on a PyeUnicam SP-1000 spectrophotometer. 1H and 13C NMR spectra were measured on a Varian EM-390-200 MHz in DMSO as solvent using TMS as internal standard, and chemical shifts are expressed as 5. Analytical data were obtained from the Microa-nalytical Data Unit at Cairo University, Giza, Egypt. General procedure for the synthesis of the imidazo [1,2-£]isoqumolines 2a-c To a solution of 1 (1.57 g, 10 mmol) in ethanol (25 mL) containing triethylamine (0.5 mL), cyclohexanone (0.98 g, 10 mmol) and any of benzaldehyde (1.06 g, 10 mmol), 4-chlorobenzaldehyde (1.40 g, 10 mmol) or 4-methoxybenzaldehyde (1.36 g, 10 mmol) were added. The reaction mixture was heated under reflux for 3 h, then poured into a beaker containing ice/water mixture containing a few drops of hydrochloric acid. The solid product formed was collected by filtration and dried. The obtained product was crystallized from ethanol to give greenish brown crystals. 11-Phenyl-7,8,9,10-tetrahydrobenzo[4,5]imidazo[1,2-¿]isoquinoline-6-carbonitrile (2a). Yield 2.26 g (70%); m.p. 223-225 °C; Anal. Calcd. for C22H17N3 (323.39): C, 81.71; H, 5.30; N, 12.99%; found: C, 81.60; 4.85; N, 13.20%. IR (KBr) v/cm-1 3092-3030 (CH aromatic), 2887 (CH-alph.), 2222 (CN), 1523 (C=N), 1589, 1437 (C=C). 1H NMR (DMSO-d6) 5 2.49-2.88 (m, 8H, 4CH2-cyclohexanone), 7.13-8.01 (m6 , 9H, C6H4, C6H5). 13C NMR (DMSO-d6) 5 38.6, 39.0, 40.2, 40.6 (4xCH2), 116.2 (CN), 120.3, 122.8, 124.9, 127.8, 128.0, 129.8, 131.2, 132.6, 134.5, 134.8, 144.3, 146.8, 150.4 (2xC6H4, pyridine C), 164.8 (C=N). MS (m/z) 323 (M+, 23%). 11-(4-Chlorophenyl)-7,8,9,10-tetrahydrobenzo[4,5] imidazo[1,2-£]isoquinolme-6-carbomtrile (2b). Yield 2.32 g (65%); m.p. 267-269 °C; Anal. Calcd. for C22H16ClN3 (357.84): C, 73.84; H, 4.51; N, 11.74%; found: C, 73.61; H, 4.38; N, 11.94%. IR (KBr) v/cm-1 3091- 3027 (CH aromatic), 2900 (CH-alph.), 2222 (CN), 1584, 1488 (C=C), 1523 (C=N). 1H NMR (DMSO-d6) 5 2.49-2.51 (m, 8H, 4CH2-cyclohexanone), 7.26-8.01 6(m, 8H, 2C6H4). 13C NMR (DMSO-d6) 5 38.8, 39.12, 39.7, 39.9 (4xCH2), 115.9 (CN), 120.0, 122.9, 125.8, 126.2, 130.2, 131.1, 134.5, 136.1, 138.4, 140.2, 144.9, 145.6, 150.3 (2xC6H4, pyridine-C), 165.2 (C=N). MS (m/z) 357 (M+, 80%). 11-(4-Methoxyphenyl)-7,8,9,10-tetrahydrobenzo[4,5] imidazo[1,2-£] isoquinoline-6-carbonitrile (2c). Yield 2.58 g (73%); m.p. 243-246 °C; Anal. Calcd. for C23H19N3O (353.42): C, 78.16; H, 5.42; N, 11.89%; found: C, 777.33; H, 4.05; N, 11.06%. IR (KBr) v/cm-1 3103-3015 (CH aromatic), 2901-2838 (CH-alph.), 2212 (CN), 1643 (C=O), 1512 (C=N), 1589, 1446 (C=C). 1H NMR (DMSO-d6) 5 1.30 (s, 3H, CH3), 2.49-2.51 (m, 8H, 4xCH2-cyclohexanone), 6.91-8.02 (m, 8H, 2C6H4). 13C NMR (DMSO-d6) 5 38.55, 38.84, 39.39, 39.67, 39.95 (4-CH2, cyclohexanone), 55.5 (CH3), 116.7 (CN), 120.3, 123.6, 124.6, 125.3, 130.8, 131.1, 134.5, 136.6, 138.8, 140.6, 143.7, 146.8, 150.2 (2xC6H4, pyridine-C), 165.8 (C=N). MS (m/z) 353 (M+, 36%). 1-Methyl-3-(phenylamino)benzo[4,5]imidazo[1,2-a] pyridine-4-carbonitrile (3). To a mixture of 1 (1.57 g, 10 mmol) and acetoaceta-nilide (1.77 g, 10 mmol), ammonium acetate (0.77 g, 10 Mohareb et al.: New Approaches for the Synthesis, Cytotoxicity ... Acta Chim. Slov. 2016, 63, 227-240 235 mmol) was added. The reaction mixture was heated in oil bath at 140 °C for 1 h then left to cool. The semisolid formed was triturated with ethanol (40 mL) and the formed solid product was collected by filtration and dried. The obtained product was crystallized from ethanol to give light amber crystals. Yield 2.27 g (76%); m.p. > 300 °C; Anal. Calcd. for C19H14N4 (298.34): C, 76.49; H, 4.73; N, 18.78%; found: C, 7(5.33; H, 4.44; N, 18.63%. IR (KBr) v/cm-1 3423-3106 (NH), 3056 (CH aromatic), 2955-2829 (CH-alph.), 2208 (CN), 1569, 1489 (C=C), 1526 (C=N). 1H NMR (DMSO-d6) 5 1.91 (s, 3H, CH3), 5.94 (s, 1H, pyridine C5), 7.05-7.55 (m, 9H, C6H4 C6H5), 13.19 (s, 1H, NH). 13C NMR (DMSO-d6) 5 20.24 (CH3), 116.2 (CN), 111.1, 115.9, 120.7, 120.76, 122.1, 126.2, 127.5, 128.7, 131.5, 104.2, 146.6, 150.8, 154.0, 155.1 (C6H4, C6H5, pyridine-C), 164.1 (C=N). MS (m/z) 298 (M+, 18%). 1-Methyl-2-(2-phenylhydrazono)-3-(phenylimino)-1,2, 3,4-tetrahydrobenzo-[4,5]imidazo-[1,2-a]pyridine-4-carbonitrile (4) To a cold (0-5 °C) solution of 3 (1.20 g, 40.2mmol) in ethanol (50 mL) containing sodium hydroxide solution (10 mL, 10%) and a solution of benzenediazonium chloride (40.2 mmol) [which was prepared by dissolving sodium nitrite (0.60 g, 80.4 mmol) in water (2 mL) was added to a cold solution of aniline (0.4 mL, 40.2 mmol) containing the appropriate amount of hydrochloric acid with continuous stirring] was added with continuous stirring. The reaction mixture was stirred at room temperature for 3 h and the solid product formed was collected by filtration and dried. The obtained product was crystallized from ethanol to give red crystals. Yield 2.67 g (66%); m.p. 245-248 °C; Anal. Calcd. for C^H^N (404.47): C, 74.24; H, 4.98; N, 20.78%; found: C, 74.05; H, 4.88; N, 19.82%. IR (KBr) v/cm-1 3419-3200 (NH), 3058 (CH aromatic), 2961-2854 (CH-alph.), 2210 (CN), 1600 (C=N), 1450 (C=C). 1H NMR (DMSO-d^ 5 1.91 (s, 3H, CH3), 5.82 (s, 1H, pyridi-ne C6), 7.17 (s,6 1H, pyridine C3),37.25-7.79 (m, 14H, QH^xQHj), 8.53 (s, 1H, NH). 13C NMR (DMSO-d^ 5 19.9 (CH3), 115.8 (CN), 118.1, 120.7, 120.7, 120.7, 121.4, 122.0, 122.9, 123.1, 123.9, 124.5, 124.9, 125.83, 126.1, 127.3, 128.7, 141.3, 156.1 (C6H4, 2xC6H5, pyridi-ne-C), 173.2, 186.9 (2xC=N). MS (m/z) 404 (M+, 66%). 2-(1-Acetyl-1_ff-benzo[d]imidazol-2(3ff)-ylidene) ace-tonitrile (5) A solution of 1 (1.57 g, 10 mmol) in acetic acid (15 mL) and acetic anhydride (35 mL) was heated under reflux till a precipitate is formed after 30 min, then poured into a beaker containing ice/water mixture. The solid product formed was collected by filtration and dried. The obtained product was crystallized from ethanol to give gold crystals. Yield 1.47 g (74%); m.p. > 300 °C; Anal. Calcd. for C11H9N3O (199.21): C, 66.32; H, 4.55; N, 21.09%; found: C, 64.48; H, 4.33; N, 21.63%. IR (KBr) v/cm-1 3069 (CH aromatic), 2965-2871 (CH-alph.), 2192 (CN), 1630 (C=O), 1594, 1410 (C=C), 1513 (C=N). 1H NMR (DM-SO-d6) 5 2.21 (s, 3H, CH3), 3.30 (s, 2H, CH2), 7.2-7.51 (m, 4H, C6H4). 13C NMR (DMSO-d6) 5 27.0 (CH3), 65.3 (CH2), 115.84 (CN), 121.1, 123.1, 124.0, 125.3, 126.0, 130.3 (C6H4), 151.1 (C=O), 189.2 (C=N). MS (m/z) 199 (M+, 49%). 2-(1-Bromo-1_ff-benzo[d]imidazol-2-yl)acetonitrile (6) A solution of 5 (1.99 g, 10 mmol) in glacial acetic acid (10 mL) was warmed to 60 oC, then bromine (0.08 g, 10 mmol) in acetic acid (10 mL) was added drop-wise with continuous stirring. The reaction mixture was stirred for 1.5 h then poured into ice/water and the solid product formed was collected by filtration. The obtained product was crystallized from ethanol to give brownish orange crystals. Yield 1.95 g (70%); m.p. > 300 °C; Anal. Calcd. for C11H8BrN3O (278.10): C, 47.51; H, 2.90; N, 15.11%; found: C, 47.33; H, 3.89; N, 13.49%. IR (KBr) v/cm-1 3050 (CH aromatic), 2971 (CH-alph.), 2193 (CN), 1630 (C=O), 1602, 1474 (C=C). 1H NMR (DMSO-d6) 5 3.30, 4.00 (2s, 4H, 2xCH2), 7.20-7.63 (m, 4H, C6H4). 63C NMR (DMSO-d6) 5 55.8, 62.7 (2xCH2), 116.46 (CN), 119.2, 121.3, 1246.8, 129.3, 133.6 (C6H4), 168.8 (C=O), 172.7 (C=N). MS (m/z) 278 (M+, 28%). 3-Amino-1-hydroxybenzo[4,5]imidazo[1,2-a]pyridine- 4-carbonitrile (7) To a solution of 6 (2.78 g, 10 mmol) in dimethylfor-mamide (5 mL) heated on a water bath at 60 °C potassium cyanide (0.65 g, 10 mmol), dissolved in a least amount of water, was added while stirring. The reaction mixture was left in the water bath for 30 min at 60 °C then poured into a beaker containing ice/water mixture and a few drops of hydrochloric acid. The solid product formed was collected by filtration and dried. The obtained product was crystallized from ethanol to give reddish brown crystals. Yield 1.93 g (86%); m.p. > 300 °C; Anal. Calcd. for C12H8N4O (224.22): C, 64.28; H, 3.60; N, 24.99%; found: C, 63.88; H, 3.88; N, 24.69%. IR (KBr) v/cm-1 3436-3233 (OH, NH2), 3050 (CH aromatic), 2193 (CN), 1513 (C=N), 1597 (C=C). 1H NMR (DMSO-d6) 5 5.21 (s, 2H, D2O exchangeable, NH2), 7.22 (s, 1H, pyridine C5), 7.21-7.88 (m, 4H, C6H4), 12.91 (s, 1H, OH). 13C NMR (DMSO-d6) 5 116.8 (CN), 119.2, 121.3, 124.8, 129.3, 133.6, 136.2, 136.8, 140.1, 142.4, 144.1, 146.7 (C6H4). MS (m/z) 224 (M+, 60%). 6 4 2-(1-(2-Hydrazinylacetyl)-1_ff-benzo[d]imidazol-2-yl) acetonitrile (8) To a solution of 6 (2.78 g, 10 mmol) in dimethylfor-mamide (5 mL) hydrazine hydrate (0.50 g, 10 mmol) was added. The reaction mixture was stirred for 3 h at room temperature then poured into a beaker containing acidi- Mohareb et al.: New Approaches for the Synthesis, Cytotoxicity ... 236 Acta Chim. Slov. 2016, 63, 227-240 fied ice/water mixture. The solid product formed was collected by filtration and dried. The obtained product was crystallized from ethanol to give light brown crystals. Yield 2.01 g (88%); m.p. > 300 °C; Anal. Calcd. for C11H11N5O (229.24): C, 57.63; H, 4.84; N, 30.55%; found: C, 57.80; H, 4.69; N, 30.79%. IR (KBr) v/cm-1 3414-3214 (NH, NH2), 3100 (CH aromatic), 2900 (CH-alph.), 2194 (CN), 1668 (C=O), 1600, 1470 (C=C), 1519 (C=N). 1H NMR (DMSO-d6) 5 3.34 (s, 2H, CH2), 4.40 (s, 2H, CH2), 7.21-7.63 (m, 4H, C6H4), 5.76 (s, 2H, NH2), 12.74 (s, 1H, NH). 13C NMR (DMSO-d6) 5 56.8, 65.9 (2xch2), 120.8, 122.4, 126.7, 128.3, 133.6, 145.9 (C6H4), 164.2 (C=O), 172.8 (C=N). MS (m/z) 229 (M+, 18%). 2-(1-(2-Aminothiazol-4-yl)-1_ff-benzo[d]imidazol-2-yl) acetonitrile (9) To a solution of 6 (2.78 g, 10 mmol) in dimethylfor-mamide (20 mL), thiourea (0.76 g, 10 mmol) was added. The reaction mixture was heated under reflux for 3 h then left to cool. The solid product formed was collected by filtration and dried. The obtained product was crystallized from ethanol to give brown crystals. Yield 2.12 g (83%); m.p. > 300 °C; Anal. Calcd. for C12H9N5S (255.30): C, 56.45; H, 3.55; N, 27.43; S, 12.56%; found: C, 56.78; H, 3.69; N, 27.39; S, 12.48%. IR (KBr) v/cm-1 3428-3227 (NH2), 3050 (CH aromatic), 2967-2922 (CH-alph.), 2195 (CN), 1513 (C=N), 1597, 1474 (C=C). 1H NMR (DMSO-d6) 5 3.30 (s, 2H, CH2), 5.21 (s, 2H, NH2), 6.23 (s, 1H, thiazole C5), 7.24-7.63 (m, 4H, C6H4). 13C NMR (DMSO-d6) 5 55.6, (CH2), 116.8 (CN), 120.8, 122.4, 126.7, 128.3, 133.6, 134.3, 138.5, 145.9 (C6H4, thiazole C), 168.2, 172.8 (2xC=N). MS (m/z) 255 (M+, 25%). General procedure for the synthesis of the thiophene derivatives 11a-c To a solution of 5 (1.99 g, 10 mmol) in dimethylfor-mamide (20 mL) containing finely divided potassium hydroxide (0.56 g, 10 mmol), phenylisothiocyanate (1.35 g, 10 mmol) was added. The reaction mixture was stirred at room temperature for 24 h, then any of chloroacetone (0.92 g, 10 mmol), ethyl chloroacetate (1.22 g, 10 mmol) or a-bromoacetophenone (1.99 g, 10 mmol) was added. The whole reaction mixture was stirred at room temperature for additional 24 h. The solid product formed upon dilution with ice/water mixture containing hydrochloric acid (till pH 6) was collected by filtration and dried. The obtained product was crystallized from ethanol to give copper-coloured crystals for 11a and 11b and brown crystals for 11c. 1-(4-(1-Acetyl-1_ff-benzo[d]imidazol-2-yl)-3-amino-5-(phenylamino)thiophen-2-yl)ethanone (11a) Yield 3.36 g (86%); m.p. > 300 °C; Anal. Calcd. For C21H18N4O2S (390.46): C, 64.60; H, 4.65; N, 14.35; S, 8.21%; found: C, 64.88; H, 4.67; N, 14.72; S, 8.40%. IR (KBr) v/cm-1 3456-3210 (NH), 3064 (CH aromatic), 2966-2875 (CH-alph.), 2192 (CN), 1730 (C=O), 1598, 1470 (C=C), 1516 (C=N). 1H NMR (DMSO-d6) 5 1.84, 1.89 (2s, 6H, 2xCH3), 5.48 (s, 2H, NH2), 7.12-7.51 (m, 9H, C6H4 C6H5), 12.71 (s, 1H, NH). 13C NMR (DMSO-d6) 5 27.0 (CH3), 38.8 (CH2), 115.8 (CN), 121.4, 123.1, 1244.9, 125.1, 126.2, 127.9, 128.2, 129.4, 130.3, 133.2, 134.8, 136.8, 140.2, 142.1 (C6H4, C6H5, thiophene C), 163.8, 166.2 (2 C=O), 182.5 (C=N). MS (m/z) 390 (M+, 38%). Ethyl 4-(1-acetyl-1_ff-benzo[d]imidazol-2-yl)-3-amino-5-(phenylamino)-thiophene-2-carboxylate (11b) Yield 2.94 g (70%); m.p. 140 °C; Anal. Calcd. for C22H20N4O3S (420.48): C, 62.84; H, 4.79; N, 13.32; S, 7.63%; found: C, 62.73; H, 4.52; N, 13.06; S, 7.67%. IR (KBr) v/cm-1 3441-3211 (NH), 3063 (CH aromatic), 2970-2876 (CH-alph.), 2193 (CN), 1725 (C=O), 1597, 1472 (C=C), 1516 (C=N). 1H NMR (DMSO-d6) 5 1.16-1.21 (t, 3H, CH3), 2.11 (s, 3H, CH3), 3.97 (s, 2H, NH2), 4.11-4.16 (q, 2H, CH2), 6.76-7.55 (m, 9H, C6H4 C6H5), 12.74 (s, 1H, NH). 13C NMR (DMSO-d6) 5 16.3 (ester CH3), 28.2 (CO-CH3), 52.8 (ester CH2), 115.9 (CN), 120.6, 122.8, 123.4, "125.0, 125.8, 126.3, 126.8, 128.1, 130.3, 133.4, 136.1, 138.9, 141.8, 142.0 (C6H4, C6H5, thiophene C), 164.3, 166.9 (2xC=O), 172.8 (C=N). M6S (5m/z) 420 (M+, 26%). 1-(2-(4-Amino-5-benzoyl-2-(phenylamino)thiophen-3-yl)-1ff-benzo[d]imidazol-1-yl)ethanone (11c) Yield 2.26 g (50%); m.p. > 300 °C; Anal. Calcd. for C26H20N4O2S (452.53): C, 69.01; H, 4.45; N, 12.38; S, 7.2069%20; f4ou2nd: C, 68.89; H, 4.65; N, 12.09; S, 6.83%. IR (KBr) v/cm-1 3431-3212 (NH, NH2), 3067 (CH aromatic), 2968-2879 (CH-alph.), 2195 (CN), 1690, 1627 (2 C=O), 1599, 1471 (C=C), 1516 (C=N). 1H NMR (DMSO-d6) 5 2.12 (s, 3H, CH3), 2.73 (s, 2H, D2O exchangeable, NH2), 7.21-8.09 (m, 14H, C6H4, 2xC6H5), 8.74 (s, 1H, D20 exchangeable, NH). 13C NMR (DMSO-d6) 5 28.6 (CO-CllJ, 119.3, 121.9, 124.2, 125.8, 125.8, 126.0, 126.8, 1227.1, 127.9, 128.1, 130.3, 133.4, 136.1, 138.9, 140.3, 141.2, 142.6, 143.8 (2xC6H5, thiophene C), 163.9, 165.2 (2xC=O), 170.6 (C=N). MS (m/z) 452 (M+, 42%). General procedure for the synthesis of the benzylidene derivatives 12a,b To a solution of 5 (1.99 g, 10 mmol) in dimethylfor-mamide (25 mL) containing piperidine (0.5 mL), either of benzaldehyde (1.06 g, 10 mmol) or salicylaldehyde (1.22 g, 10 mmol) was added. The reaction mixture was heated under reflux for 3 h then poured into a beaker containing ice/water mixture containing a few drops of hydrochloric acid. The solid product formed was collected by filtration and dried. The obtained product was crystallized from et-hanol to give yellow crystals of 12a and orange crystals of 12b. Mohareb et al.: New Approaches for the Synthesis, Cytotoxicity ... Acta Chim. Slov. 2016, 63, 227-240 237 2-(1-Acetyl-1_ff-benzo[d]imidazol-2-yl)-3-phenylacry-lonitrile (12a) Yield 1.81 g (63%); m.p. > 300 °C; Anal. Calcd. for C18H13N3O (287.32): C, 75.25; H, 4.56; N, 14.63%; found: C, 74.33; H, 4.36; N, 14.49%. IR (KBr) v/cm-1 3066 (CH aromatic), 2967-2876 (CH-alph.), 2193 (CN), 1627 (C=O), 1599, 1470 (C=C), 1515 (C=N). 1H NMR (DMSO-d6) 5 2.00 (s, 3H, CH3), 3.30 (s, 1H, CH), 7.21-7.51 (m, 9H, C6H4 C6H5). 13C NMR (DMSO-d6) 5 28.4 (CO-CllJ, 88.5, 9Ci.6 (CH=C), 121.6, 123.8, 124.2, 124.9, 125.03, 127.1, 130.3, 133.4, 138.9, 139.0 (2xC6H5), 165.8 (C=O), 171.2 (C=N). MS (m/z) 287 (M+, 23%). 2-(1-Acetyl-1ff-benzo[d]imidazol-2-yl)-3-(2-hydroxyp-henyl)acrylonitrile (12b) Yield 2.79 g (92%); m.p. > 300 °C; Anal. Calcd. for C18H13N3O2 (303.31): C, 71.28; H, 4.32; N, 13.85%; found: C, 71.33; H, 4.88; N, 9.04%. IR (KBr) v/cm-1 3432-3215 (OH), 3100 (CH aromatic), 2999-2882 (CH-alph.), 2194 (CN), 1633 (C=O), 1601, 1477 (C=C), 1518 (C=N). 1H NMR (DMSO-d6) 5 1.20 (s, 3H, CH3), 3.57 (s, 1H, CH), 7.21-7.51 (m, 8H, 2C6H4), 12.75 (s, 1H, D2O exchangeable, OH). 13C NMR (DMSO-d6) 5 28.6 (CO-CH3), 88.9, 90.8 (CH=C), 120.8, 122.5, 124.2, 124.6, 125.(5, 126.8, 128.7, 133.4, 136.3, 140.3 (2xC6H5), 164.6 (C=O), 170.8 (C=N). MS (m/z) 303 (M+, 36%). General procedure for the synthesis of the ben-zo[4,5]imidazo[1,2-a]pyridine derivatives 13a,b To a solution of 5 (1.99 g, 10 mmol) in dimethylfor-mamide (30 mL) containing triethylamine (0.5 mL), either malononitrile (0.66 g, 10 mmol) or ethyl cyanoacetate (1.13 g, 10 mmol) was added. The reaction mixture was heated under reflux for 4 h then poured into a beaker containing ice/water mixture containing a few drops of hydrochloric acid. The solid product formed was collected by filtration and dried. The obtained product was crystallized from ethanol to give buff crystals of 13a and yellowish brown crystals of 13b. 3-Amino-1-methylbenzo[4,5]imidazo[1,2-a]pyridine-2,4-dicarbonitrile (13a) Yield 2.04 g (82%); m.p. > 300 °C; Anal. Calcd. For C14H11N5 (249.27): C, 67.46; H, 4.45; N, 28.10%; found: C, 67.06; H, 4.18; N, 28.43%. IR (KBr) v/cm-1 3470-3212 (NH, NH2), 3077 (CH aromatic), 2971-2877 (CH-alph.), 2223, 2194 (2CN), 1599, 1474 (C=C), 1517 (C=N). 1H NMR (DMSO-d6) 5 1.16 (s, 3H, CH3), 5.51 (s, 2H, D2O exchangeable, NH2), 6.30 (s, 1H, pyridine C6), 7.21-7.50 (m, 4H, C6H4), 12.75 (s, 1H, NH). 13C NMR (DMSO-d6) 5 22.7 (CH3), 116.8, 117.3 (2xCN), 119.8, 120.7, 1226.5, 124.8, 125.3, 126.4, 128.0, 130.9, 133.2, 134.8, 143.2 (C6H4, imidazole, pyridine C), 173.8 (C=N). MS (m/z) 249 (M+, 22%). Ethyl 3-amino-4-cyano-1 -methylbenzo[4,5]imidazo [1,2-a]pyridine-2-carboxylate (13b) Yield 2.01 g (68%); m.p. > 300 °C; Anal. Calcd. for C16H16N4O2 (296.32): C, 64.85; H, 5.44; N, 18.91%; found6 C, 64.92; H, 4.27; N, 18.73%. IR (KBr) v/cm-1 3426-3214 (NH, NH2), 3103 (CH aromatic), 2883 (CH-alph.), 2194 (CN), 1750 (C=O, ester), 1601, 1472 (C=C). 1H NMR (DMSO-d6) 5 1.10 (t, 3H, CH3), 2.12 (s, 3H, CH3), 3.29 (q, 2H, CH2), 5.01 (s, 2H, D20 exchangeable, NH2), 6.10 (s, 1H, pyridine C6), 7.20-7.51 (m, 4H, C6H4), 12.773 (s, 1H, D2O exchangeable, NH). 13C NMR (DMSO-d6) 5 16.82 (ester CH3), 22.4 (CH3), 56.9 (ester CH2), 116.5, 117.1 (2xcn), 119.9, 121.3, 122.8, 124.4, 125.1, 126.8, 127.4, 130.3, 133.6, 134.1, 143.8 (C6H4, imidazole, pyridine C), 162.8 (C=O). MS (m/z) 296 (M+, 35%). 2-(1-Acetyl-1_ff-benzo[d]imidazol-2-yl)-3-phenylbut-2-enenitrile (14) To a mixture of 5 (1.99 g, 10 mmol) and acetophe-none (1.35 g, 10 mmol), ammonium acetate (0.77 g, 10 mmol) was added. The reaction mixture was heated in oil bath at 140 °C for 1 h then left to cool. The semisolid formed was triturated with ethanol (40 mL) and the formed solid product was collected by filtration and dried. The obtained product was crystallized from ethanol to give dark brown crystals. Yield 1.99 g (66%); m.p. 210-213 °C; Anal. Calcd. for C19H15N3O (301.34): C, 75.73; H, 5.02; N, 13.94%; found: C, 74.83; H, 5.39; N, 13.85%. IR (KBr) v/cm-1 3086 (CH aromatic), 2880 (CH-alph.), 2194 (CN), 1630 (C=O), 1599, 1472 (C=C), 1517 (C=N). 1H NMR (DMSO-d6) 5 1.20, 2.40 (2s, 6H, 2xCH3), 7.21-7.52 (m, 9H, C6H4 C6H5). 13C NMR (DMSO-d6) 5 22.4, 28.6 (CH3, CO-CH3)', 88.9, 90.6 (CH=C), 121.3, 122.5, 124.2, 124.65, 126.1, 126.8, 127.2, 133.4, 136.3, 136.9 (2xc6h5), 164.6 (C=O), 170.6 (C=N). MS (m/z) 301 (M+, 17%). 2-(1-Acetyl-1_ff-benzo[d]imidazol-2-yl)-3,5-diphenyl-penta-2,4-dienenitrile (15) To a solution of 14 (3.01 g, 10 mmol) in dimethyl-formamide (25 mL) containing piperidine (0.50 mL), benzaldehyde (1.06 g, 10 mmol) was added. The reaction mixture was heated under reflux for 4 h then poured into a beaker containing ice/water mixture containing a few drops of hydrochloric acid. The solid product formed was collected by filtration and dried. The obtained product was crystallized from ethanol to give brown crystals. Yield 2.57 g (66%); m.p. 150 °C; Anal. Calcd. for C26H19N3O (389.45): C, 80.18; H, 4.92; N, 10.79%; found: C, 80.29; H, 5.14; N, 10.59%. IR (KBr) v/cm-1 3057 (CH aromatic), 2927 (CH-alph.), 2195 (CN), 1627 (C=O), 1596 (C=N), 1489 (C=C). 1H NMR (DMSO-d6) 5 2.23 (s, 3H, CH3), 2.72, 2.89 (2s, 2H, 2xCH), 6.50-8.35 (m, 14H, C6H4 2xC6H5). 13C NMR (DMSO-d6) 5 24.3 (CH3), 98.5, 104.8 6C=C), 116.4 (CN), 120.4, 121.3, 122.4, 124.9, 125.8, 126.8, 127.8, 128.0, 130.6, 133.5, Mohareb et al.: New Approaches for the Synthesis, Cytotoxicity ... 238 Acta Chim. Slov. 2016, 63, 227-240 136.5, 140.8 (C6H4, 2xC6H5), 164.3 (C=O), 173.1 (C=N). MS (m/z) 389 (M+, 44%). General procedure for the synthesis of the thiophene derivatives 16a,b To a solution of 5 (1.99 g, 10 mmol) in 1,4-dioxane (25 mL) containing triethylamine (0.50 mL) and elemental sulfur (0.32 g, 10 mmol) either malononitrile (0.66 g, 10 mmol) or ethyl cyanoacetate (1.13 g, 10 mmol) was added. The reaction mixture was heated under reflux for 4 h then poured into a beaker containing ice/water mixture containing a few drops of hydrochloric acid. The solid product was collected by filtration and dried. The obtained product was crystallized from ethanol to give dark brown crystals of 16a and light brown crystals of 16b. 5-(1-Acetyl-1_ff-benzo[d]imidazol-2-yl)-2,4-diaminot-hiophene-3-carbonitrile (16a) Yield 2.08 g (70%); m.p. 288-293 °C; Anal. Calcd. for C14H11N5OS (297.34): C, 56.55; H, 3.73; N, 23.55; S, 10.78%; found: C, 56.43; H, 3.49; N, 23.70; S, 10.63%. IR (KBr) v/cm-1 3426-3213 (2xNH2), 3050 (CH aromatic), 2973, 2882 (CH-alph.), 2195 (CN), 1680 (C=O), 1600 (C=N), 1470 (C=C), 1518 (C=N). 1H NMR (DMSO-d6) 5 2.06 (s, 3H, CH3), 2.73, 2.89 (2s, 4H, D2O exchangeable, 2xNH2), 7.20-73.95 (m, 4H, C6H4). 13C NMR (DMSO-d6) 5 22.6 (CH3), 116.3 (CN), 119.8, 120.7, 122.6, 124.6, 129.5, 130.4, 133.5, 142.8, 143.2, 144.3 (C6H4, thiophene C), 163.2 (C=O), 172.3 (C=N). MS (m/z) 297 (M+, 32%). Ethyl 5-(1-acetyl-1_ff-benzo[d]imidazol-2-yl)-2,4-dia-minothiophene-3-carboxylate (16b) Yield 2.38 g (69%); m.p. > 300 °C; Anal. Calcd. for C16H16N4O3S (344.39): C, 55.80; H, 4.68; N, 16.27; S, 9.31%; found: C, 55.89, H, 4.29; N, 16.60; S, 8.04%. IR (KBr) v/cm-1 3430-3216 (2 NH2), 3107 (CH aromatic), 2978, 2884 (CH-alph.), 1633 (C=O), 1750 (C=O, ester), 1601 (C=N), 1471 (C=C), 1518 (C=N). 1H NMR (DMSO-d6) 5 1.04 (t, 3H, CH3), 2.33 (s, 3H, CH3), 4.28 (q, 2H, CH2), 5.80, 5.95 (2s, 4H, D2O exchangeable, 2 NH2), 7.21-7.51 (m, 4H, C6H4). 13C NMR (DMSO-d6) 5 16.22 (ester CH3), 22.8 (CH3), 120.3, 120.8, 121.2, 123.8, 124.8, 125.8, 134.8, 142.8, 143.6, 144.9 (C6H4, thiophene C), 164.8 (C=O), 172.0 (C=N). MS (m/z) 344 (M+, 48%). 2-(2-Phenylhydrazono)-2-(1-acetyl-1_ff-benzo[d]imida-zol-2-yl) acetonitrile (17) To a cold solution (0-5 oC) of 5 (1.99 g, 10 mmol) in ethanol (50 mL) containing sodium hydroxide solution (10 mL, 10%) and a solution of benzenediazonium chloride (10 mmol) [which was prepared by dissolving sodium nitrite (0.70 g, 10 mmol) in water, 2 mL was added to a cold solution of aniline (0.93 g, 10 mmol) containing appropriate amount of hydrochloric acid and with continuous stirring] was added with continuous stirring. The solid product formed was collected by filtration and dried. The obtained product was crystallized from ethanol to give brown crystals. Yield 2.24 g (74%); m.p. 265 °C; Anal. Calcd for C17H13N5O (303.32): C, 67.32; H, 4.32; N, 23.09%; found: C, 67.29; H, 4.09; N, 23.27%. IR (KBr) v/cm-1 3423-3214 (NH), 3098 (CH aromatic), 2976-2883 (CH-alph.), 2194 (CN), 1631 (C=O), 1521 (=N-NH), 1601, 1475 (C=C). 1H NMR (DMSO-d6) 5 2.21 (s, 3H, CH3), 7.21-7.51 (m, 9H, C6H4, C6H5), 12.76 (s, 1H, D2O exchangeable, NH). 13C NMR (DMSO-d6) 5 22.8 (CH3), 115.9 (CN), 120.4, 120.7, 123.2, 124.6, 126.5, 128.6, 133.5, 142.8, 143.2, 144.3 (C6H5, C6H4), 164.0 (C=O), 172.8 (C=N). MS (m/z) 303 (M+, 20%). General procedure for the synthesis of the phenylhy-drazone derivatives 18a,b To a solution of 17 (3.03 g, 10 mmol) in 1,4-dioxane (30 mL) containing triethylamine (0.50 mL) and elemental sulfur (0.32 g, 10 mmol) either malononitrile (0.66 g, 10 mmol) or ethyl cyanoacetae (1.13 g, 10 mmol) was added. The reaction mixture was heated under reflux for 4 h then poured into a beaker containing ice/water mixture containing a few drops of hydrochloric acid. The solid product was collected by filtration and dried. The obtained product was crystallized from ethanol to give yellow crystals of 18a and light green crystals of 18b. 4-(2-((2-Phenylhydrazono)(cyano)methyl)-1-ff-benzo [d]imidazol-1-yl)-2-aminothiophene-3-carbonitrile (18a) Yield 3.45 g (90%); m.p. 198 °C; Anal. Calcd. for C20H13N7S (383.43): C, 62.65; H, 3.42; N, 25.57; S, 8.36%; found: C, 62.88; H, 3.59; N, 25.19; S, 8.07%. IR (KBr) v/cm-1 3352-3209 (NH, NH2), 3100 (CH-aroma-tic), 2922 (CH-alph.), 2196 (CN), 1547 (C=N), 1463 (C=C). 1H NMR (DMSO-d6) 5 5.60 (s, 2H, D2O exchangeable, NH2), 7.20 (s, 1H, thiophene C5), 7.21-7.67 (m, 9H, C6H4 C6H5), 12.75 (s, 1H, NH). 13C NMR (DMSO-d6) 5 116.3. 116.8 (2xCN), 120.0, 120.6, 123.2, 125.1, 126.5, 127.2, 136.5, 138.1, 139.2, 140.6, 140.8, 141.3, 142.6 (C6H5, C6H4, thiophene C), 164.0 (C=O), 172.8 (C=N). MS 5m/z) 383 (M+, 18%). Ethyl 4-(2-((2-phenylhydrazono)(cyano)methyl)-1_ff-benzo[d]imidazol-1-yl)-2-aminothiophene-3-carboxy-late (18b) Yield 2.54 g (59%); m.p. > 300 °C; Anal. Calcd. for C22H18N6O2S (430.48): C, 61.38; H, 4.21; N, 19.52; S, 7.2425%18; f6ou2nd: C, 61.03; H, 4.58; N, 19.71; S, 7.66%. IR (KBr) v/cm-1 3427-3215 (NH, NH2), 3100 (CH aromatic), 2900 (CH-alph.), 2194 (CN), 1632 (C=O), 1520 (C=N), 1601, 1475 (C=C). 1H NMR (DMSO-d6) 5 1.10 (t, 3H, CH3), 4.21 (q, 2H, CH2), 5.01 (s, 2H, D2O exchangeable, NH2), 7.20 (s, 1H, thiophene C5), 7.21-7.51 (m, 9H, C6H4, C6H5), 12.77 (s,1H, D2O exchangeable, NH). 13C NMR (I6MSO-d6) 5 16.3 (ester CH3), 54.8 (ester Mohareb et al.: New Approaches for the Synthesis, Cytotoxicity ... Acta Chim. Slov. 2016, 63, 227-240 239 CH2), 116.5 (CN), 120.3, 121.8, 123.8, 125.1, 127.2, 128.6, 130.2, 138.1, 139.2, 139.9, 140.4, 142.9, 143.8 (C6H5, C6H4, thiophene C), 164.0 (C=O), 172.8 (C=N). MS (m/z) 430 (M+, 22%). 5. Conclusions In the present study we have synthesized a series of heterocyclic derivatives of 2-cyanomethylbenzo[c]imida-zole 1. The newly synthesized products were tested against MCF-7 (breast adenocarcinoma), NCI-H460 (non-small cell lung cancer), and SF-268 (CNS cancer) and the results showed that compounds 2b, 6, 11b, 11c, 12b, 16a, 16b and 18a exhibited optimal cytotoxic effect against cancer cell lines. The toxicity of these optimal cytotoxic compounds was monitored via in vivo lethality to shrimp larvae (Arte-mia salina). The results showed that compounds 11b, 12b, and 16b are non toxic towards shrimp larvae. 6. References 1. H. Irannejad, M. Amini, F. Khodagholi, N. Ansari, S. K. Tu-si, M. Sharifzadeh, A. Shafiee, Bioorg. Med. Chem. 2010, 18, 4224-4230. http://dx.doi.org/10.1016/j.bmc.2010.04.097 2. N. Catozzi, M. G. Edwards, S. A. Raw, P. Wasnaire, R. K. Taylor, J. Org. Chem. 2009, 74, 8343-8354. http://dx.doi.org/10.1021/jo901761r 3. S. Laphookhieo, S. Jones, S. A. Raw, Y. F. Sainz, R. J. K. Taylor, Tetrahedron Lett. 2006, 47, 3865-3870. http://dx.doi.org/10.1016/j.tetlet.2006.03.178 4. D. L. Boger, J. S. Panek, M. Yasuda, Org. Synth. 1988, 66, 142-150. http://dx.doi.org/10.15227/orgsyn.066.0142 5. R. M. Mohareb, K. El-Sharkawy, S. M. Sherif, Acta Pharm. 2008, 58, 429-444. 6. A. M. Salah, Heteroatom Chem. 2003, 14, 612-614. http://dx.doi.org/10.1002/hc.10199 7. P. A. Crooks, L. P. Dwoskin, Biochemical Pharmacology 1997, 54, 743-753. http://dx.doi.org/10.1016/S0006-2952(97)00117-2 8. B. K. Warren, E. E. Knaus, Eur. J. Med. Chem. 1987, 22, 411-415. http://dx.doi.org/10.1016/0223-5234(87)90028-6 9. B. J. Mulchin, C. G. Newton, J. W. Baty, C. H. Grasso, W. J. Martin, M. C. Walton, E. M. Dangerfield, C. H. Plunkett, M. V. Berridge, J. L. Harper, M. S. Timmer, B. L. Stocker, Bioorg. Med. Chem. 2010, 18, 3238-3251. http://dx.doi.org/10.1016/j.bmc.2010.03.021 10. Y.-L. Luo, K. Baathulaa, V. K. Kannekanti, C.-H. Zhou, G.-X. Cai, Sci. China Chem. 2015, 58, 483-494. http://dx.doi.org/10.1007/s11426-014-5296-3 11. J. N. Sangshetti, P. P. Dharmadhikari, R. S. Chouthe, B. Fa-tema, V. Lad, V. Karande, S. N. Darandale, D. B. Shinde, Bi-oorg. Med. Chem. Lett. 2013, 23, 2250-2253. http://dx.doi.Org/10.1016/j.bmcl.2013.01.041 12. J. Witherington, V. Bordas, S. L. Garland, D. M. B. Hickey, R. J. Ife, J. Liddle, M. Saunders, D. G. Smith, R. W. Ward, Bioorg. Med. Chem. Lett. 2003, 13, 1577-1580. http://dx.doi.org/10.1016/S0960-894X(03)00134-3 13. X.-H. Liu, H.-F. Liu, X. Shen, B.-A. Song, P. S. Bhadury, H.-L. Zhu, J.-X. Liu, X.-B. Qi, Bioorg. Med. Chem. Lett. 2010, 20, 4163-4167. http://dx.doi.org/10.1016/j.bmcl.2010.05.080 14. S. Ram, Progress in Medicinal Chemistry 1988, 25, 233-247. http://dx.doi.org/10.1016/S0079-6468(08)70279-0 15. M. Gaba, S. Singh, C. Mohan, Eur. J. Med. Chem. 2014, 76, 494-505. http://dx.doi.org/10.1016Zj.ejmech.2014.01.030 16. Y. M. Shaker, M. A. Omar, K. Mahmoud, S. M. Elhallouty, W. M. El-Senousy, M. M. Ali, A. E. Mahmoud, A. H. Abdel-Halim, S. M. Soliman, H. I. El Diwani, J. Enzyme Inhib. Med. Chem. 2015, 8, 1-20. 17. J. J. B. Nevado, G. C. Penalvo, R. M. R. Dorado, V. R. Robledo, J. Pharm. Biomed. Anal. 2014, 92, 211-219. http://dx.doi.org/10.1016/jjpba.2013.12.020 18. S. Fujimoto, Eur. J. Pharm. 1994, 265, 159-166. http://dx.doi.org/10.1016/0014-2999(94)90426-X 19. A. D. Khoje, C. Charnock, B. Wan, S. Franzblau, L.-L. Gun-dersen, Bioorg. Med. Chem. 2011, 19, 3483-3491. http://dx.doi.org/10.1016/j.bmc.2011.04.023 20. D. Seenaiah, P. R. Reddy, G. M. Reddy, A. Padmaja, V. Pad-mavathi, N. S. Krishna, Eur. J. Med. Chem. 2014, 77, 1-7. http://dx.doi.org/10.1016/j.ejmech.2014.02.050 21. E. C. Herrmann, J. A. Herrmann, D. C. Delong, Antiviral Research, 1981, 1, 301-314. http://dx.doi.org/10.1016/0166-3542(81)90022-X 22. S. Sharma, S. Gangal, A. Rauf, Eur. J. Med. Chem. 2009, 44, 1751-1757. http://dx.doi.org/10.1016/j.ejmech.2008.03.026 23. M. Rashid, A. Husain, R. Mishra, Eur. J. Med. Chem. 2012, 54, 855-866. http://dx.doi.org/10.1016/j.ejmech.2012.04.027 24. R. S. Harapanhalli, R. W. Howell, D. V. Rao, Nuclear Med. Biol. 1994, 21, 641-647. http://dx.doi.org/10.1016/0969-8051(94)90030-2 25. S. Singh, B. S. Dwarakanath, T. L. Mathew, J. Phtotochem. Photobiol. B. 2004, 77, 45-54. http://dx.doi.org/10.1016/S1011-1344(04)00122-8 26. H. A. S. Elzahabi, Eur. J. Med. Chem. 2011, 46, 4025-4034. http://dx.doi.org/10.1016/j.ejmech.2011.05.075 27. J. R. Tseng, D. Stuart, K. Aardalen, A. Kaplan, N. Aziz, N. P. Hughes, S. S. Gambhir, Neoplasia 2011, 13, 266-275. http://dx.doi.org/10.1593/neo.101466 28. T. R. Reddy, G. R. Reddy, L. S. Reddy, S. Jammula, Y. Lin-gappa, R. Kapavarapu, C. L. Meda, K. V. Parsa, M. Pal, Eur. J. Med. Chem. 2012, 48, 265-274. http://dx.doi.org/10.1016/j.ejmech.2011.12.024 29. S. Khaksar, M. Yaghoobi, J. Fluor. Chem. 2012, 142, 41-44. http://dx.doi.org/10.1016/jjfluchem.2012.06.009 30. R. M. Mohareb, S. M. Sherif, Arch. Die Pharm. (Weinheim), 1991, 324, 469-471. http://dx.doi.org/10.1002/ardp.2503240801 Mohareb et al.: New Approaches for the Synthesis, Cytotoxicity ... 240 Acta Chim. Slov. 2016, 63, 227-240 31. R. M. Mohareb, N. I. Abdel-Sayed, S. M. Sherif, Phosphorus, Sulfur, Silicon Relat. Elem. 1991, 63, 119-129. http://dx.doi.org/10.1080/10426509108029435 32. R. M. Mohareb, Monatsh. Chem. 1992, 123, 341-347. http://dx.doi.org/10.1007/BF00810946 33. J. Chodosh, R. D. Dix, R. C. Howell, W. G. Stroop, S. C. G. Tseng, Invest. Ophthalmol. Vis. Sci. 1994, 35, 1046-1058. 34. A. U. Rahman, M. I. Choudhary, W. J. Thomsen, Bioassay Techniques for Drug Development, Harwood Academic, Amsterdam, The Netherlands, 2001. http://dx.doi.org/10.4324/9780203304532 35. B. Brayn, M. Timothy, S. Tore, General and Applied Toxicology, 2nd ed., vol. I, 52 p. 36. J. L. Carballo, Z. L. H. Inda, P. Pérez, M. D. G. Grávalos, BMC Biotechnol. 2002, 2, 17-22. http://dx.doi.org/10.1186/1472-6750-2-17 37. M. Calleja, G. Persoone, Atla. 1992, 20, 396-405. 38. P. N. Chatterjee, S. Roy, Tetrahedron 2011, 67, 4569-4577. http://dx.doi.org/10.1016Zj.tet.2011.04.092 Povzetek Pri reakciji etil cianoacetat z o-fenilendiaminom je nastal 2-cianometilbenzo[c]imidazol (1). To spojino smo uporabili kot ključno izhodno snov za sintezo biološko aktivnih heterocikličnih derivatov. Pri reakciji 1 s cikloheksanonom ter benzaldehidom, 4-metoksibenzaldehidom oz. 4-klorobenzaldehidom so nastali pripojeni derivati 2a-c. Testiranje anti-tumorske aktivnosti novopripravljenih produktov proti trem rakastim celičnim linijam, t.j. MCF-7 (adeno-carcinom dojke), NCI-H460 (nemikrocelični karcinom pljuč) in SF-268 (rak centralnega živčnega sistema), je pokazalo, da spojine 2b, 6, 11b, 11c, 12b, 16a, 16b in 18a kažejo optimalno citotoksično učinkovitost proti tem rakastim linjam z IC50 vrednostmi v nM območju. Ker so bioaktivne spojine pogosto strupene za ličinke morskih rakcev, smo se odločili preveriti še in vivo strupenost teh spojin na ličinke Artemia salina. Spojine 11b, 12b in 16b niso pokazale nobene strupenosti za testirane organizme. Mohareb et al.: New Approaches for the Synthesis, Cytotoxicity ...